Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Cell Rep ; 42(6): 112625, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37294634

ABSTRACT

Endogenous retroviruses (ERVs) have rewired host gene networks. To explore the origins of co-option, we employed an active murine ERV, IAPEz, and an embryonic stem cell (ESC) to neural progenitor cell (NPC) differentiation model. Transcriptional silencing via TRIM28 maps to a 190 bp sequence encoding the intracisternal A-type particle (IAP) signal peptide, which confers retrotransposition activity. A subset of "escapee" IAPs (∼15%) exhibits significant genetic divergence from this sequence. Canonical repressed IAPs succumb to a previously undocumented demarcation by H3K9me3 and H3K27me3 in NPCs. Escapee IAPs, in contrast, evade repression in both cell types, resulting in their transcriptional derepression, particularly in NPCs. We validate the enhancer function of a 47 bp sequence within the U3 region of the long terminal repeat (LTR) and show that escapee IAPs convey an activating effect on nearby neural genes. In sum, co-opted ERVs stem from genetic escapees that have lost vital sequences required for both TRIM28 restriction and autonomous retrotransposition.


Subject(s)
Endogenous Retroviruses , Tripartite Motif-Containing Protein 28 , Animals , Mice , Cell Differentiation , Embryonic Stem Cells/metabolism , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Histones/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Terminal Repeat Sequences/genetics
2.
Front Cell Dev Biol ; 10: 941493, 2022.
Article in English | MEDLINE | ID: mdl-36172281

ABSTRACT

Rett syndrome is a human intellectual disability disorder that is associated with mutations in the X-linked MECP2 gene. The epigenetic reader MeCP2 binds to methylated cytosines on the DNA and regulates chromatin organization. We have shown previously that MECP2 Rett syndrome missense mutations are impaired in chromatin binding and heterochromatin reorganization. Here, we performed a proteomics analysis of post-translational modifications of MeCP2 isolated from adult mouse brain. We show that MeCP2 carries various post-translational modifications, among them phosphorylation on S80 and S421, which lead to minor changes in either heterochromatin binding kinetics or clustering. We found that MeCP2 is (di)methylated on several arginines and that this modification alters heterochromatin organization. Interestingly, we identified the Rett syndrome mutation site R106 as a dimethylation site. In addition, co-expression of protein arginine methyltransferases (PRMT)1 and PRMT6 lead to a decrease of heterochromatin clustering. Altogether, we identified and validated novel modifications of MeCP2 in the brain and show that these can modulate its ability to bind as well as reorganize heterochromatin, which may play a role in the pathology of Rett syndrome.

3.
J Insect Physiol ; 116: 57-69, 2019 07.
Article in English | MEDLINE | ID: mdl-31039373

ABSTRACT

Neural development depends on the controlled proliferation and differentiation of neural precursors. In holometabolous insects, these processes must be coordinated during larval and pupal development. Recently, protein arginine methylation has come into focus as an important mechanism of controlling neural stem cell proliferation and differentiation in mammals. Whether a similar mechanism is at work in insects is unknown. We investigated this possibility by determining the expression pattern of three protein arginine methyltransferase mRNAs (PRMT1, 4 and 5) in the developing brain of bumblebees by in situ hybridisation. We detected expression in neural precursors and neurons in functionally important brain areas throughout development. We found markedly higher expression of PRMT1, but not PRMT4 and PRMT5, in regions of mushroom bodies containing dividing cells during pupal stages at the time of active neurogenesis within this brain area. At later stages of development, PRMT1 expression levels were found to be uniform and did not correlate with actively dividing cells. Our study suggests a role for PRMT1 in regulating neural precursor divisions in the mushroom bodies of bumblebees during the period of neurogenesis.


Subject(s)
Bees/genetics , Gene Expression , Insect Proteins/genetics , Mushroom Bodies/growth & development , Neurogenesis/physiology , Protein-Arginine N-Methyltransferases/genetics , Animals , Bees/growth & development , Bees/metabolism , Brain/growth & development , Insect Proteins/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Pupa/genetics , Pupa/growth & development , Pupa/metabolism , RNA, Messenger/metabolism , Time Factors
4.
Exp Eye Res ; 159: 114-122, 2017 06.
Article in English | MEDLINE | ID: mdl-28228349

ABSTRACT

Corneal endothelial cells (CECs) are essential for maintaining corneal stromal hydration and ensuring its transparency, which is necessary for normal vision. Dysfunction of CECs leads to stromal decompensation, loss of transparency and corneal blindness. Corneal endothelium has low proliferative potential compared to surface epithelial cells leading to poor regeneration of CEC following injury. Additionally, the tissue exhibits age related decline in endothelial cell density with re-organisation of the cell layer, but no regeneration. The mechanisms which control proliferation and differentiation of neural crest derived CEC progenitors are yet to be clearly elucidated. Prdm (Positive regulatory domain) family of transcriptional regulators and chromatin modifiers are important for driving differentiation of a variety of cellular types. Many Prdm proteins are expressed in specific precursor cell populations and are necessary for their progression to a fully differentiated phenotype. In the present work, we sought to identify members of the Prdm gene family which are specifically expressed in human (h) CECs with a view to begin addressing their potential roles in CEC biology, focussing especially on Prdm 4 and 5 genes. By performing semi-quantitative reverse transcription coupled to PCR amplification we found that in addition to Prdm4 and Prdm5, Prdm2 and Prdm10 genes are expressed in hCECs. We further found that cultured primary hCECs or immortalised HCEC-12 cells express all of the Prdm genes found in CECs, but also express additional Prdm transcripts. This difference is most pronounced between Prdm gene expression patterns of CECs isolated from healthy human corneas and immortalised HCEC-12 cells. We further investigated Prdm 4 and Prdm 5 protein expression in cultured primary hCECs and HCEC-12 cells as well as in a human cadaveric whole cornea. Both Prdm 4 and Prdm 5 are expressed in human corneal endothelium, primary hCECs and in HCECs-12 cells, characterised by expression of the Na+/K+-ATPase. We observed that both proteins exhibit cytosolic (intracellular, but non-nuclear and distinct from extracellular fluid) as well as nuclear localisation within the endothelial layer, with Prdm 5 being more concentrated in the nuclei of the endothelial cells than Prdm 4. Thus, our work identifies novel Prdm genes specifically expressed in corneal endothelial cells which may be important in the control of CEC differentiation and proliferation.


Subject(s)
Corneal Diseases/genetics , DNA-Binding Proteins/genetics , Endothelium, Corneal/metabolism , Gene Expression Regulation , RNA/genetics , Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Corneal Diseases/metabolism , Corneal Diseases/pathology , DNA-Binding Proteins/biosynthesis , Endothelium, Corneal/pathology , Humans , Immunohistochemistry , Microscopy, Confocal , Regeneration/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis
5.
EMBO J ; 36(5): 604-616, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28122869

ABSTRACT

An unresolved question is how HIV-1 achieves efficient replication in terminally differentiated macrophages despite the restriction factor SAMHD1. We reveal inducible changes in expression of cell cycle-associated proteins including MCM2 and cyclins A, E, D1/D3 in macrophages, without evidence for DNA synthesis or mitosis. These changes are induced by activation of the Raf/MEK/ERK kinase cascade, culminating in upregulation of CDK1 with subsequent SAMHD1 T592 phosphorylation and deactivation of its antiviral activity. HIV infection is limited to these G1-like phase macrophages at the single-cell level. Depletion of SAMHD1 in macrophages decouples the association between infection and expression of cell cycle-associated proteins, with terminally differentiated macrophages becoming highly susceptible to HIV-1. We observe both embryo-derived and monocyte-derived tissue-resident macrophages in a G1-like phase at frequencies approaching 20%, suggesting how macrophages sustain HIV-1 replication in vivo Finally, we reveal a SAMHD1-dependent antiretroviral activity of histone deacetylase inhibitors acting via p53 activation. These data provide a basis for host-directed therapeutic approaches aimed at limiting HIV-1 burden in macrophages that may contribute to curative interventions.


Subject(s)
G1 Phase , HIV-1/physiology , Immune Evasion , Macrophages/immunology , Macrophages/virology , Monomeric GTP-Binding Proteins/metabolism , Protein Processing, Post-Translational , Cells, Cultured , HIV-1/immunology , Humans , Immunity, Innate , Phosphorylation , SAM Domain and HD Domain-Containing Protein 1
6.
Biochem Biophys Res Commun ; 474(2): 388-394, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27125459

ABSTRACT

PRDM4 is a member of the PRDM family of transcriptional regulators which control various aspects of cellular differentiation and proliferation. PRDM proteins exert their biological functions both in the cytosol and the nucleus of cells. All PRDM proteins are characterised by the presence of two distinct structural motifs, the PR/SET domain and the zinc finger (ZF) motifs. We previously observed that deletion of all six zinc fingers found in PRDM4 leads to its accumulation in the cytosol, whereas overexpressed full length PRDM4 is found predominantly in the nucleus. Here, we investigated the requirements for single zinc fingers in the nuclear localisation of PRDM4. We demonstrate that ZF's 1, 2, 5 and 6 contribute to the accumulation of PRDM4 in the nucleus. Their effect is additive as deleting either ZF1-2 or ZF 5-6 redistributes PRDM4 protein from being almost exclusively nuclear to cytosolic and nuclear. We investigated the potential mechanism of nuclear shuttling of PRDM4 via the importin α/ß-mediated pathway and find that PRDM4 nuclear targeting is independent of α/ß-mediated nuclear import.


Subject(s)
Cell Nucleus/metabolism , Neural Stem Cells/metabolism , Subcellular Fractions/metabolism , Zinc Fingers/physiology , Active Transport, Cell Nucleus/physiology , Animals , HEK293 Cells , Humans , Mice , PC12 Cells , Rats , Signal Transduction/physiology
7.
Neurosci Lett ; 550: 87-92, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23831350

ABSTRACT

During neuronal development, the neuroepithelial stem cells (NSCs) initially undergo proliferative divisions, later switching to neurogenic ones whereby one NSC and a post-mitotic neuron are generated. We recently showed that a member of the PRDM family of transcriptional regulators, PRDM4/SC1, recruits a type II protein arginine methyltransferase, PRMT5, to maintain the "stem-like" cellular state of the embryonic mouse cortical NSCs. However, little is known about the regulation of activity of this complex under proliferation- or differentiation-inducing growth conditions. In the present work I investigate the regulation of SC1/PRMT5-mediated methylation activity in PC12 cells treated with EGF or NGF. I present evidence that NGF down-regulates SC1/PRMT5 methyltransferase (MTase) activity and that the reduction in SC1/PRMT5 MTase activity occurs mainly in the nucleus. I suggest that high levels of SC1/PRMT5 activity are associated with the proliferative state of the cells.


Subject(s)
DNA-Binding Proteins/genetics , Epidermal Growth Factor/pharmacology , Nerve Growth Factor/pharmacology , Neurons/drug effects , Protein Methyltransferases/genetics , Transcription Factors/genetics , Animals , Arginine/metabolism , Cell Proliferation/drug effects , Cells, Cultured , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Methylation , Mice , Neurons/metabolism , PC12 Cells , Protein Methyltransferases/metabolism , Rats , Transcription Factors/metabolism
8.
Curr Biol ; 22(19): R835-8, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-23058801

ABSTRACT

Social insects represent a unique model for how the same genome can give rise to entirely different phenotypes - soldiers, common labourers, and queens. New research on ants and honeybees points to DNA methylation as a crucial factor in determining the caste of a developing individual.


Subject(s)
Ants/genetics , DNA Methylation , Gene Expression Regulation , Genome, Insect , Animals
9.
J Biol Chem ; 287(51): 42995-3006, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23048031

ABSTRACT

During development of the cerebral cortex, neural stem cells (NSCs) undergo a temporal switch from proliferative (symmetric) to neuron-generating (asymmetric) divisions. We investigated the role of Schwann cell factor 1 (SC1/PRDM4), a member of the PRDM family of transcription factors, in this critical transition. We discovered that SC1 recruits the chromatin modifier PRMT5, an arginine methyltransferase that catalyzes symmetric dimethylation of histone H4 arginine 3 (H4R3me2s) and that this modification is preferentially associated with undifferentiated cortical NSCs. Overexpressing SC1 in embryonic NSCs led to an increase in the number of Nestin-expressing precursors; mutational analysis of SC1 showed that this was dependent on recruitment of PRMT5. We found that SC1 protein levels are down-regulated at the onset of neurogenesis and that experimental knockdown of SC1 in primary NSCs triggers precocious neuronal differentiation. We propose that SC1 and PRMT5 are components of an epigenetic regulatory complex that maintains the "stem-like" cellular state of the NSC by preserving their proliferative capacity and modulating their cell cycle progression. Our findings provide evidence that histone arginine methylation regulates NSC differentiation.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Histones/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Protein Methyltransferases/metabolism , Transcription Factors/metabolism , Animals , Arginine , Cell Proliferation , Cerebral Cortex/cytology , Cerebral Cortex/embryology , DNA-Binding Proteins/chemistry , Embryo, Mammalian/cytology , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunoprecipitation , Methylation , Mice , PC12 Cells , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases , RNA, Small Interfering/metabolism , Rats , Time Factors , Transcription Factors/chemistry
10.
PLoS One ; 5(11): e13807, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21072194

ABSTRACT

BACKGROUND: Epigenetic modifications regulate key transitions in cell fate during development of the central nervous system (CNS). During cortical development the initial population of proliferative neuroepithelial precursor cells give rise to neurons and then glia in a strict temporal order. Neurogenesis and gliogenesis are accompanied by a switch from symmetric to asymmetric divisions of the neural precursor cells generating another precursor and a differentiated progeny. To investigate whether specific post-translational histone modifications define specific stages of neural precursor differentiation during cortical development I focussed on the appearance of two different types of histone arginine methylation, the dimethyl symmetric H4R3 (H4R3me2s) and dimethyl asymmetric H4R3 (H4R3me2a) in the developing mouse cortex. METHODOLOGY/PRINCIPAL FINDINGS: An immunohistochemical study of the developing cortex at different developmental stages was performed to detect the distribution of H4R3me2s and H4R3me2a modifications. I analysed the distribution of these modifications in: 1) undifferentiated neural precursors, 2) post-mitotic neurons and 3) developing oligodendrocyte precursors (OLPs) using lineage-specific and histone modification-specific antibodies to co-label the cells. I found that the proliferative neuroepithelium during the stage of mainly symmetric expansive divisions is characterised by the prevalence of H4R3me2s modification and almost no detectable H4R3me2a modification. However, at a later stage, when the cortical layers with post-mitotic neurons have begun forming, both H4R3me2a and H4R3me2s modifications are detected in the post-mitotic neurons and in the developing OLPs. CONCLUSIONS/SIGNIFICANCE: I propose that the H4R3me2s modification forms part of the "histone code" of undifferentiated neural precursors. The later appearance of the H4R3me2a modifications specifies the onset of neurogenesis and gliogenesis and the commitment of the NSCs to differentiate. Thus, the sequential appearance of the two different H4R3 methylation marks may define a particular cellular state of the NSCs during their development and differentiation demonstrating the role of histone arginine methylation in cortical development.


Subject(s)
Arginine/metabolism , Cerebral Cortex/metabolism , Histones/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Epithelium/embryology , Epithelium/metabolism , Fluorescent Antibody Technique , Methylation , Mice , Neurogenesis , Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors
12.
J Endocrinol ; 198(1): 185-91, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18463147

ABSTRACT

Although the zonation of the adrenal cortex has a clear functional role, the mechanisms that maintain it remain largely conjectural. The concept that an outer proliferative layer gives rise to cells that migrate inwards, adopting sequentially the zona glomerulosa, fasciculata and reticularis phenotypes, has yet to be explained mechanistically. In other tissues, Eph receptor (EphR)/ephrin signalling provides a mechanism for cellular orientation and migration patterns. Real-time PCR and other methods were used to determine the possible role of Eph/ephrin systems in the rat adrenal. mRNA coding for several members of the EphR family was detected, but out of these, EphA2 provided the closest parallel to zonal organisation. In situ hybridisation showed that EphA2 mRNA and EphA protein were predominantly located in the zona glomerulosa. Its transcription closely reflected expected changes in the glomerulosa phenotype, thus it was increased after a low-sodium diet, but decreased by pretreatment with the angiotensin-converting enzyme inhibitor, captopril. It was also decreased by ACTH treatment, but unaffected by betamethasone. mRNA coding for ephrin A1, the major ligand for the EphA receptors, was also detected in the rat adrenal, though changes evoked by the various pretreatments did not clearly reflect the expected changes in zonal function. Because the maintenance of cellular zonation requires clear positional signals within the adrenal cortex, these data support a role for Eph forward and reverse signalling in the maintenance of adrenocortical zonation.


Subject(s)
Adrenal Cortex/metabolism , Receptors, Eph Family/genetics , Adrenocorticotropic Hormone/pharmacology , Animals , Cytochrome P-450 CYP11B2/physiology , Ephrins/genetics , Male , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptor, EphA2/genetics , Receptors, Eph Family/physiology
13.
Proc Natl Acad Sci U S A ; 103(20): 7871-6, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16684879

ABSTRACT

Ciliary neurotrophic factor (Cntf) plays an essential role in postnatal maintenance of spinal motoneurons. Whereas the expression of this neurotrophic factor is low during embryonic development, it is highly up-regulated after birth in myelinating Schwann cells of rodents. To characterize the underlying transcriptional mechanisms, we have analyzed and compared the effects of various glial transcription factors. In contrast to Pit-1, Oct-1, Unc-86 homology region (POU) domain class 3, transcription factor 1 (Oct6/SCIP/Tst-1) and paired box gene 3 (Pax3), SRY-box-containing gene 10 (Sox10) induces Cntf expression in Schwann cells. Subsequent promoter analysis using luciferase reporter gene and EMSA identified the corresponding response elements within the Cntf promoter. Overexpression of Sox10 in primary sciatic nerve Schwann cells leads to a >100-fold up-regulation of Cntf protein, and suppression of Sox10 by RNA interference in the spontaneously immortalized Schwann cell line 32 reduces Cntf expression by >80%. Mice with heterozygous inactivation of the Sox10 gene show significantly reduced Cntf protein levels in sciatic nerves, indicating that Sox10 is necessary and sufficient for regulating Cntf expression in the peripheral nervous system.


Subject(s)
Ciliary Neurotrophic Factor , DNA-Binding Proteins/metabolism , Gene Expression Regulation , High Mobility Group Proteins/metabolism , Schwann Cells/physiology , Transcription Factors/metabolism , Animals , Binding Sites , Cell Line , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , DNA-Binding Proteins/genetics , Genes, Reporter , High Mobility Group Proteins/genetics , Mice , Promoter Regions, Genetic , Rats , SOXE Transcription Factors , Schwann Cells/cytology , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Transcription Factors/genetics , Transcription Initiation Site
14.
J Cell Biol ; 164(7): 985-96, 2004 Mar 29.
Article in English | MEDLINE | ID: mdl-15051733

ABSTRACT

Schwann cell factor 1 (SC1), a p75 neurotrophin receptor-interacting protein, is a member of the positive regulatory/suppressor of variegation, enhancer of zeste, trithorax (PR/SET) domain-containing zinc finger protein family, and it has been shown to be regulated by serum and neurotrophins. SC1 shows a differential cytoplasmic and nuclear distribution, and its presence in the nucleus correlates strongly with the absence of bromodeoxyuridine (BrdU) in these nuclei. Here, we investigated potential transcriptional activities of SC1 and analyzed the function of its various domains. We show that SC1 acts as a transcriptional repressor when it is tethered to Gal4 DNA-binding domain. The repressive activity requires a trichostatin A-sensitive histone deacetylase (HDAC) activity, and SC1 is found in a complex with HDACs 1, 2, and 3. Transcriptional repression exerted by SC1 requires the presence of its zinc finger domains and the PR domain. Additionally, these two domains are involved in the efficient block of BrdU incorporation by SC1. The zinc finger domains are also necessary to direct SC1's nuclear localization. Lastly, SC1 represses the promoter of a promitotic gene, cyclin E, suggesting a mechanism for how growth arrest is regulated by SC1.


Subject(s)
Cell Cycle/physiology , Cyclin E/genetics , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Repressor Proteins/metabolism , 3T3 Cells , Animals , Calcium-Binding Proteins , Cell Cycle/genetics , Cell Line , Extracellular Matrix Proteins , Humans , Mice , PC12 Cells , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Receptor, Nerve Growth Factor , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/genetics , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL